1
|
Devasia AJ, Chari A, Lancman G. Bispecific antibodies in the treatment of multiple myeloma. Blood Cancer J 2024; 14:158. [PMID: 39266530 PMCID: PMC11393350 DOI: 10.1038/s41408-024-01139-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/24/2024] [Revised: 08/29/2024] [Accepted: 09/02/2024] [Indexed: 09/14/2024] Open
Abstract
The treatment paradigm in myeloma is constantly changing. Upfront use of monoclonal antibodies like daratumumab along with proteasome inhibitors (PI)s, and immune modulators (IMiD)s have significantly improved survival and outcomes, but also cause unique challenges at the time of relapse. Engaging immune T cells for tumour cell kill with chimeric antigenic T-cell (CAR T-cell) therapy and bispecific antibodies have become important therapeutic options in relapsed multiple myeloma. Bispecific antibodies are dual antigen targeting constructs that engage the T cells to plasma cells through various target antigens like B-cell membrane antigen (BCMA), G-protein-coupled receptor family C group 5 member D (GPRC5D), and Fc receptor-homolog 5 (FcRH5). These agents have proven to induce deep and durable responses in heavily pre-treated myeloma patients with a predictable safety profile and the ease of off-the-shelf availability. Significant research is ongoing to overcome resistance mechanisms like T cell exhaustion, target antigen mutation or loss and high disease burden. Various trials are also studying these agents as first line options in the newly diagnosed setting. These agents play an important role in the relapsed setting, and efforts are underway to optimize their sequencing in the myeloma treatment algorithm.
Collapse
Affiliation(s)
| | - Ajai Chari
- University of California, San Francisco, San Francisco, CA, USA
| | - Guido Lancman
- Princess Margaret Cancer Centre, Toronto, ON, Canada.
| |
Collapse
|
2
|
Schoenfeld K, Harwardt J, Kolmar H. Better safe than sorry: dual targeting antibodies for cancer immunotherapy. Biol Chem 2024; 405:443-459. [PMID: 38297991 DOI: 10.1515/hsz-2023-0329] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/17/2023] [Accepted: 01/11/2024] [Indexed: 02/02/2024]
Abstract
Antibody-based therapies are revolutionizing cancer treatment and experience a steady increase from preclinical and clinical pipelines to market share. While the clinical success of monoclonal antibodies is frequently limited by low response rates, treatment resistance and various other factors, multispecific antibodies open up new prospects by addressing tumor complexity as well as immune response actuation potently improving safety and efficacy. Novel antibody approaches involve simultaneous binding of two antigens on one cell implying increased specificity and reduced tumor escape for dual tumor-associated antigen targeting and enhanced and durable cytotoxic effects for dual immune cell-related antigen targeting. This article reviews antibody and cell-based therapeutics for oncology with intrinsic dual targeting of either tumor cells or immune cells. As revealed in various preclinical studies and clinical trials, dual targeting molecules are promising candidates constituting the next generation of antibody drugs for fighting cancer.
Collapse
Affiliation(s)
- Katrin Schoenfeld
- Institute for Organic Chemistry and Biochemistry, Technical University of Darmstadt, Peter-Grünberg-Strasse 4, D-64287 Darmstadt, Germany
| | - Julia Harwardt
- Institute for Organic Chemistry and Biochemistry, Technical University of Darmstadt, Peter-Grünberg-Strasse 4, D-64287 Darmstadt, Germany
| | - Harald Kolmar
- Institute for Organic Chemistry and Biochemistry, Technical University of Darmstadt, Peter-Grünberg-Strasse 4, D-64287 Darmstadt, Germany
- Centre for Synthetic Biology, Technical University of Darmstadt, Darmstadt, Germany
| |
Collapse
|
3
|
Grab AL, Kim PS, John L, Bisht K, Wang H, Baumann A, Van de Velde H, Sarkar I, Shome D, Reichert P, Manta C, Gryzik S, Reijmers RM, Weinhold N, Raab MS. Pre-Clinical Assessment of SAR442257, a CD38/CD3xCD28 Trispecific T Cell Engager in Treatment of Relapsed/Refractory Multiple Myeloma. Cells 2024; 13:879. [PMID: 38786100 PMCID: PMC11120574 DOI: 10.3390/cells13100879] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/02/2024] [Revised: 05/02/2024] [Accepted: 05/10/2024] [Indexed: 05/25/2024] Open
Abstract
Current treatment strategies for multiple myeloma (MM) are highly effective, but most patients develop relapsed/refractory disease (RRMM). The anti-CD38/CD3xCD28 trispecific antibody SAR442257 targets CD38 and CD28 on MM cells and co-stimulates CD3 and CD28 on T cells (TCs). We evaluated different key aspects such as MM cells and T cells avidity interaction, tumor killing, and biomarkers for drug potency in three distinct cohorts of RRMM patients. We found that a significantly higher proportion of RRMM patients (86%) exhibited aberrant co-expression of CD28 compared to newly diagnosed MM (NDMM) patients (19%). Furthermore, SAR442257 mediated significantly higher TC activation, resulting in enhanced MM killing compared to bispecific functional knockout controls for all relapse cohorts (Pearson's r = 0.7). Finally, patients refractory to anti-CD38 therapy had higher levels of TGF-β (up to 20-fold) compared to other cohorts. This can limit the activity of SAR442257. Vactoserib, a TGF-β inhibitor, was able to mitigate this effect and restore sensitivity to SAR442257 in these experiments. In conclusion, SAR442257 has high potential for enhancing TC cytotoxicity by co-targeting CD38 and CD28 on MM and CD3/CD28 on T cells.
Collapse
Affiliation(s)
- Anna Luise Grab
- Heidelberg Myeloma Center, Department of Medicine V, Medical Faculty Heidelberg and University Hospital, Heidelberg University, Im Neuenheimer Feld 410, 69120 Heidelberg, Germany; (A.L.G.); (C.M.); (S.G.); (N.W.)
- Clinical Cooperation Unit Molecular Hematology/Oncology, German Cancer Research Center (DKFZ), 69120 Heidelberg, Germany
| | - Peter S. Kim
- Sanofi Research and Development, Sanofi North America, Cambridge, MA 02141, USA (K.B.); (H.W.); (H.V.d.V.)
| | - Lukas John
- Heidelberg Myeloma Center, Department of Medicine V, Medical Faculty Heidelberg and University Hospital, Heidelberg University, Im Neuenheimer Feld 410, 69120 Heidelberg, Germany; (A.L.G.); (C.M.); (S.G.); (N.W.)
- Clinical Cooperation Unit Molecular Hematology/Oncology, German Cancer Research Center (DKFZ), 69120 Heidelberg, Germany
| | - Kamlesh Bisht
- Sanofi Research and Development, Sanofi North America, Cambridge, MA 02141, USA (K.B.); (H.W.); (H.V.d.V.)
| | - Hongfang Wang
- Sanofi Research and Development, Sanofi North America, Cambridge, MA 02141, USA (K.B.); (H.W.); (H.V.d.V.)
| | - Anja Baumann
- Heidelberg Myeloma Center, Department of Medicine V, Medical Faculty Heidelberg and University Hospital, Heidelberg University, Im Neuenheimer Feld 410, 69120 Heidelberg, Germany; (A.L.G.); (C.M.); (S.G.); (N.W.)
- Clinical Cooperation Unit Molecular Hematology/Oncology, German Cancer Research Center (DKFZ), 69120 Heidelberg, Germany
| | - Helgi Van de Velde
- Sanofi Research and Development, Sanofi North America, Cambridge, MA 02141, USA (K.B.); (H.W.); (H.V.d.V.)
| | - Irene Sarkar
- LUMICKS, 1059 CM Amsterdam, The Netherlands; (I.S.); (D.S.); (R.M.R.)
| | - Debarati Shome
- LUMICKS, 1059 CM Amsterdam, The Netherlands; (I.S.); (D.S.); (R.M.R.)
| | - Philipp Reichert
- GMMG Central Study Lab, Biobank, University Hospital Heidelberg, 69120 Heidelberg, Germany;
| | - Calin Manta
- Heidelberg Myeloma Center, Department of Medicine V, Medical Faculty Heidelberg and University Hospital, Heidelberg University, Im Neuenheimer Feld 410, 69120 Heidelberg, Germany; (A.L.G.); (C.M.); (S.G.); (N.W.)
| | - Stefanie Gryzik
- Heidelberg Myeloma Center, Department of Medicine V, Medical Faculty Heidelberg and University Hospital, Heidelberg University, Im Neuenheimer Feld 410, 69120 Heidelberg, Germany; (A.L.G.); (C.M.); (S.G.); (N.W.)
| | | | - Niels Weinhold
- Heidelberg Myeloma Center, Department of Medicine V, Medical Faculty Heidelberg and University Hospital, Heidelberg University, Im Neuenheimer Feld 410, 69120 Heidelberg, Germany; (A.L.G.); (C.M.); (S.G.); (N.W.)
- Clinical Cooperation Unit Molecular Hematology/Oncology, German Cancer Research Center (DKFZ), 69120 Heidelberg, Germany
| | - Marc S. Raab
- Heidelberg Myeloma Center, Department of Medicine V, Medical Faculty Heidelberg and University Hospital, Heidelberg University, Im Neuenheimer Feld 410, 69120 Heidelberg, Germany; (A.L.G.); (C.M.); (S.G.); (N.W.)
- Clinical Cooperation Unit Molecular Hematology/Oncology, German Cancer Research Center (DKFZ), 69120 Heidelberg, Germany
| |
Collapse
|
4
|
Klein C, Brinkmann U, Reichert JM, Kontermann RE. The present and future of bispecific antibodies for cancer therapy. Nat Rev Drug Discov 2024; 23:301-319. [PMID: 38448606 DOI: 10.1038/s41573-024-00896-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 01/22/2024] [Indexed: 03/08/2024]
Abstract
Bispecific antibodies (bsAbs) enable novel mechanisms of action and/or therapeutic applications that cannot be achieved using conventional IgG-based antibodies. Consequently, development of these molecules has garnered substantial interest in the past decade and, as of the end of 2023, 14 bsAbs have been approved: 11 for the treatment of cancer and 3 for non-oncology indications. bsAbs are available in different formats, address different targets and mediate anticancer function via different molecular mechanisms. Here, we provide an overview of recent developments in the field of bsAbs for cancer therapy. We focus on bsAbs that are approved or in clinical development, including bsAb-mediated dual modulators of signalling pathways, tumour-targeted receptor agonists, bsAb-drug conjugates, bispecific T cell, natural killer cell and innate immune cell engagers, and bispecific checkpoint inhibitors and co-stimulators. Finally, we provide an outlook into next-generation bsAbs in earlier stages of development, including trispecifics, bsAb prodrugs, bsAbs that induce degradation of tumour targets and bsAbs acting as cytokine mimetics.
Collapse
Affiliation(s)
- Christian Klein
- Roche Pharma Research and Early Development, Roche Innovation Center Zurich, Schlieren, Switzerland.
| | - Ulrich Brinkmann
- Roche Pharma Research and Early Development, Roche Innovation Center Munich, Penzberg, Germany
| | | | - Roland E Kontermann
- Institute of Cell Biology and Immunology, University Stuttgart, Stuttgart, Germany.
| |
Collapse
|
5
|
Ulitzka M, Harwardt J, Lipinski B, Tran H, Hock B, Kolmar H. Potent Apoptosis Induction by a Novel Trispecific B7-H3xCD16xTIGIT 2+1 Common Light Chain Natural Killer Cell Engager. Molecules 2024; 29:1140. [PMID: 38474651 DOI: 10.3390/molecules29051140] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2023] [Revised: 02/28/2024] [Accepted: 02/29/2024] [Indexed: 03/14/2024] Open
Abstract
Valued for their ability to rapidly kill multiple tumor cells in succession as well as their favorable safety profile, NK cells are of increasing interest in the field of immunotherapy. As their cytotoxic activity is controlled by a complex network of activating and inhibiting receptors, they offer a wide range of possible antigens to modulate their function by antibodies. In this work, we utilized our established common light chain (cLC)-based yeast surface display (YSD) screening procedure to isolate novel B7-H3 and TIGIT binding monoclonal antibodies. The chicken-derived antibodies showed single- to low-double-digit nanomolar affinities and were combined with a previously published CD16-binding Fab in a 2+1 format to generate a potent NK engaging molecule. In a straightforward, easily adjustable apoptosis assay, the construct B7-H3xCD16xTIGIT showed potent apoptosis induction in cancer cells. These results showcase the potential of the TIGIT NK checkpoint in combination with activating receptors to achieve increased cytotoxic activity.
Collapse
Affiliation(s)
- Michael Ulitzka
- Institute for Organic Chemistry and Biochemistry, Technical University of Darmstadt, Peter-Grünberg-Str. 4, 64287 Darmstadt, Germany
| | - Julia Harwardt
- Institute for Organic Chemistry and Biochemistry, Technical University of Darmstadt, Peter-Grünberg-Str. 4, 64287 Darmstadt, Germany
| | - Britta Lipinski
- Institute for Organic Chemistry and Biochemistry, Technical University of Darmstadt, Peter-Grünberg-Str. 4, 64287 Darmstadt, Germany
| | - Hue Tran
- Institute for Organic Chemistry and Biochemistry, Technical University of Darmstadt, Peter-Grünberg-Str. 4, 64287 Darmstadt, Germany
| | - Björn Hock
- Institute for Organic Chemistry and Biochemistry, Technical University of Darmstadt, Peter-Grünberg-Str. 4, 64287 Darmstadt, Germany
| | - Harald Kolmar
- Institute for Organic Chemistry and Biochemistry, Technical University of Darmstadt, Peter-Grünberg-Str. 4, 64287 Darmstadt, Germany
- Centre of Synthetic Biology, Technical University of Darmstadt, 64283 Darmstadt, Germany
| |
Collapse
|
6
|
Mathias-Machado MC, de Jesus VHF, Jácome A, Donadio MD, Aruquipa MPS, Fogacci J, Cunha RG, da Silva LM, Peixoto RD. Claudin 18.2 as a New Biomarker in Gastric Cancer-What Should We Know? Cancers (Basel) 2024; 16:679. [PMID: 38339430 PMCID: PMC10854563 DOI: 10.3390/cancers16030679] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/20/2023] [Revised: 01/25/2024] [Accepted: 01/29/2024] [Indexed: 02/12/2024] Open
Abstract
Gastric cancer (GC) remains a formidable global health challenge, ranking among the top-five causes of cancer-related deaths worldwide. The majority of patients face advanced stages at diagnosis, with a mere 6% five-year survival rate. First-line treatment for metastatic GC typically involves a fluoropyrimidine and platinum agent combination; yet, predictive molecular markers have proven elusive. This review navigates the evolving landscape of GC biomarkers, with a specific focus on Claudin 18.2 (CLDN18.2) as an emerging and promising target. Recent phase III trials have unveiled the efficacy of Zolbetuximab, a CLDN18.2-targeting antibody, in combination with oxaliplatin-based chemotherapy for CLDN18.2-positive metastatic GC. As this novel therapeutic avenue unfolds, understanding the nuanced decision making regarding the selection of anti-CLDN18.2 therapies over other targeted agents in metastatic GC becomes crucial. This manuscript reviews the evolving role of CLDN18.2 as a biomarker in GC and explores the current status of CLDN18.2-targeting agents in clinical development. The aim is to provide concise insights into the potential of CLDN18.2 as a therapeutic target and guide future clinical decisions in the management of metastatic GC.
Collapse
Affiliation(s)
- Maria Cecília Mathias-Machado
- Division of Gastrointestinal Medical Oncology, Oncoclínicas, São Paulo 04538-132, Brazil; (M.D.D.); (M.P.S.A.); (R.D.P.)
| | | | - Alexandre Jácome
- Division of Gastrointestinal Medical Oncology, Oncoclínicas, Belo Horizonte 30360-680, Brazil;
| | - Mauro Daniel Donadio
- Division of Gastrointestinal Medical Oncology, Oncoclínicas, São Paulo 04538-132, Brazil; (M.D.D.); (M.P.S.A.); (R.D.P.)
| | | | - João Fogacci
- Division of Gastrointestinal Medical Oncology, Oncoclínicas, Rio de Janeiro 22775-003, Brazil;
| | - Renato Guerino Cunha
- Cellular Therapy Program, Division of Hematology, Oncoclínicas, São Paulo 04538-132, Brazil;
| | | | - Renata D’Alpino Peixoto
- Division of Gastrointestinal Medical Oncology, Oncoclínicas, São Paulo 04538-132, Brazil; (M.D.D.); (M.P.S.A.); (R.D.P.)
| |
Collapse
|
7
|
Giang KA, Boxaspen T, Diao Y, Nilvebrant J, Kosugi-Kanaya M, Kanaya M, Krokeide SZ, Lehmann F, Svensson Gelius S, Malmberg KJ, Nygren PÅ. Affibody-based hBCMA x CD16 dual engagers for NK cell-mediated killing of multiple myeloma cells. N Biotechnol 2023; 77:139-148. [PMID: 37673373 DOI: 10.1016/j.nbt.2023.09.002] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/04/2023] [Revised: 08/16/2023] [Accepted: 09/03/2023] [Indexed: 09/08/2023]
Abstract
We describe the development and characterization of the (to date) smallest Natural Killer (NK) cell re-directing human B Cell Maturation Antigen (hBCMA) x CD16 dual engagers for potential treatment of multiple myeloma, based on combinations of small 58 amino acid, non-immunoglobulin, affibody affinity proteins. Affibody molecules to human CD16a were selected from a combinatorial library by phage display resulting in the identification of three unique binders with affinities (KD) for CD16a in the range of 100 nM-3 µM. The affibody exhibiting the highest affinity demonstrated insensitivity towards the CD16a allotype (158F/V) and did not interfere with IgG (Fc) binding to CD16a. For the construction of hBCMA x CD16 dual engagers, different CD16a binding arms, including bi-paratopic affibody combinations, were genetically fused to a high-affinity hBCMA-specific affibody. Such 15-23 kDa dual engager constructs showed simultaneous hBCMA and CD16a binding ability and could efficiently activate resting primary NK cells and trigger specific lysis of a panel of hBCMA-positive multiple myeloma cell lines. Hence, we report a novel class of uniquely small NK cell engagers with specific binding properties and potent functional profiles.
Collapse
Affiliation(s)
- Kim Anh Giang
- Department of Protein Science, Div. Protein Engineering, AlbaNova University Center, KTH Royal Institute of Technology, S-114 21 Stockholm, Sweden
| | - Thorstein Boxaspen
- Department of Cancer Immunology, Institute for Cancer Research, Oslo University Hospital, N-0424 Oslo, Norway; Precision Immunotherapy Alliance, Institute of Clinical Medicine, University of Oslo, N-0313 Oslo, Norway
| | - Yumei Diao
- Oncopeptides AB, S-171 48 Stockholm, Sweden
| | - Johan Nilvebrant
- Department of Protein Science, Div. Protein Engineering, AlbaNova University Center, KTH Royal Institute of Technology, S-114 21 Stockholm, Sweden
| | - Mizuha Kosugi-Kanaya
- Department of Cancer Immunology, Institute for Cancer Research, Oslo University Hospital, N-0424 Oslo, Norway; Precision Immunotherapy Alliance, Institute of Clinical Medicine, University of Oslo, N-0313 Oslo, Norway
| | - Minoru Kanaya
- Department of Cancer Immunology, Institute for Cancer Research, Oslo University Hospital, N-0424 Oslo, Norway; Precision Immunotherapy Alliance, Institute of Clinical Medicine, University of Oslo, N-0313 Oslo, Norway
| | - Silje Zandstra Krokeide
- Department of Cancer Immunology, Institute for Cancer Research, Oslo University Hospital, N-0424 Oslo, Norway; Precision Immunotherapy Alliance, Institute of Clinical Medicine, University of Oslo, N-0313 Oslo, Norway
| | | | | | - Karl-Johan Malmberg
- Department of Cancer Immunology, Institute for Cancer Research, Oslo University Hospital, N-0424 Oslo, Norway; Precision Immunotherapy Alliance, Institute of Clinical Medicine, University of Oslo, N-0313 Oslo, Norway.
| | - Per-Åke Nygren
- Department of Protein Science, Div. Protein Engineering, AlbaNova University Center, KTH Royal Institute of Technology, S-114 21 Stockholm, Sweden; Science For Life Laboratory, S-171 65 Solna, Sweden.
| |
Collapse
|
8
|
Zhang M, Lam KP, Xu S. Natural Killer Cell Engagers (NKCEs): a new frontier in cancer immunotherapy. Front Immunol 2023; 14:1207276. [PMID: 37638058 PMCID: PMC10450036 DOI: 10.3389/fimmu.2023.1207276] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/17/2023] [Accepted: 07/03/2023] [Indexed: 08/29/2023] Open
Abstract
Natural Killer (NK) cells are a type of innate lymphoid cells that play a crucial role in immunity by killing virally infected or tumor cells and secreting cytokines and chemokines. NK cell-mediated immunotherapy has emerged as a promising approach for cancer treatment due to its safety and effectiveness. NK cell engagers (NKCEs), such as BiKE (bispecific killer cell engager) or TriKE (trispecific killer cell engager), are a novel class of antibody-based therapeutics that exhibit several advantages over other cancer immunotherapies harnessing NK cells. By bridging NK and tumor cells, NKCEs activate NK cells and lead to tumor cell lysis. A growing number of NKCEs are currently undergoing development, with some already in clinical trials. However, there is a need for more comprehensive studies to determine how the molecular design of NKCEs affects their functionality and manufacturability, which are crucial for their development as off-the-shelf drugs for cancer treatment. In this review, we summarize current knowledge on NKCE development and discuss critical factors required for the production of effective NKCEs.
Collapse
Affiliation(s)
- Minchuan Zhang
- Singapore Immunology Network, Agency for Science, Technology, and Research, Singapore, Singapore
| | - Kong-Peng Lam
- Singapore Immunology Network, Agency for Science, Technology, and Research, Singapore, Singapore
- Department of Microbiology and Immunology, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, Singapore
- School of Biological Sciences, College of Science, Nanyang Technological University, Singapore, Singapore
| | - Shengli Xu
- Singapore Immunology Network, Agency for Science, Technology, and Research, Singapore, Singapore
- Department of Physiology, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, Singapore
| |
Collapse
|
9
|
Park DH, Liaw K, Bhojnagarwala P, Zhu X, Choi J, Ali AR, Bordoloi D, Gary EN, O’Connell RP, Kulkarni A, Guimet D, Smith T, Perales-Puchalt A, Patel A, Weiner DB. Multivalent in vivo delivery of DNA-encoded bispecific T cell engagers effectively controls heterogeneous GBM tumors and mitigates immune escape. Mol Ther Oncolytics 2023; 28:249-263. [PMID: 36915911 PMCID: PMC10006507 DOI: 10.1016/j.omto.2023.02.004] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/04/2023] [Accepted: 02/10/2023] [Indexed: 02/18/2023] Open
Abstract
Glioblastoma multiforme (GBM) is among the most difficult cancers to treat with a 5-year survival rate less than 5%. An immunotherapeutic vaccine approach targeting GBM-specific antigen, EGFRvIII, previously demonstrated important clinical impact. However, immune escape variants were reported in the trial, suggesting that multivalent approaches targeting GBM-associated antigens may be of importance. Here we focused on multivalent in vivo delivery of synthetic DNA-encoded bispecific T cell engagers (DBTEs) targeting two GBM-associated antigens, EGFRvIII and HER2. We designed and optimized an EGFRvIII-DBTE that induced T cell-mediated cytotoxicity against EGFRvIII-expressing tumor cells. In vivo delivery in a single administration of EGFRvIII-DBTE resulted in durable expression over several months in NSG mice and potent tumor control and clearance in both peripheral and orthotopic animal models of GBM. Next, we combined delivery of EGFRvIII-DBTEs with an HER2-targeting DBTE to treat heterogeneous GBM tumors. In vivo delivery of dual DBTEs targeting these two GBM-associated antigens exhibited enhanced tumor control and clearance in a heterogeneous orthotopic GBM challenge, while treatment with single-target DBTE ultimately allowed for tumor escape. These studies support that combined delivery of DBTEs, targeting both EGFRvIII and HER2, can potentially improve outcomes of GBM immunotherapy, and such multivalent approaches deserve additional study.
Collapse
Affiliation(s)
- Daniel H. Park
- Perelman School of Medicine, The University of Pennsylvania, Philadelphia, PA, USA
- Vaccine and Immunotherapy Center, The Wistar Institute, Philadelphia, PA, USA
| | - Kevin Liaw
- Vaccine and Immunotherapy Center, The Wistar Institute, Philadelphia, PA, USA
| | | | - Xizhou Zhu
- Vaccine and Immunotherapy Center, The Wistar Institute, Philadelphia, PA, USA
| | - Jihae Choi
- Vaccine and Immunotherapy Center, The Wistar Institute, Philadelphia, PA, USA
| | - Ali R. Ali
- Vaccine and Immunotherapy Center, The Wistar Institute, Philadelphia, PA, USA
| | - Devivasha Bordoloi
- Vaccine and Immunotherapy Center, The Wistar Institute, Philadelphia, PA, USA
| | - Ebony N. Gary
- Vaccine and Immunotherapy Center, The Wistar Institute, Philadelphia, PA, USA
| | - Ryan P. O’Connell
- Perelman School of Medicine, The University of Pennsylvania, Philadelphia, PA, USA
- Vaccine and Immunotherapy Center, The Wistar Institute, Philadelphia, PA, USA
| | - Abhijeet Kulkarni
- Vaccine and Immunotherapy Center, The Wistar Institute, Philadelphia, PA, USA
| | - Diana Guimet
- Inovio Pharmaceuticals, Plymouth Meeting, PA 19462, USA
| | - Trevor Smith
- Inovio Pharmaceuticals, Plymouth Meeting, PA 19462, USA
| | | | - Ami Patel
- Vaccine and Immunotherapy Center, The Wistar Institute, Philadelphia, PA, USA
| | - David B. Weiner
- Perelman School of Medicine, The University of Pennsylvania, Philadelphia, PA, USA
- Vaccine and Immunotherapy Center, The Wistar Institute, Philadelphia, PA, USA
| |
Collapse
|
10
|
Ho M, Xiao A, Yi D, Zanwar S, Bianchi G. Treating Multiple Myeloma in the Context of the Bone Marrow Microenvironment. Curr Oncol 2022; 29:8975-9005. [PMID: 36421358 PMCID: PMC9689284 DOI: 10.3390/curroncol29110705] [Citation(s) in RCA: 11] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/18/2022] [Revised: 11/08/2022] [Accepted: 11/18/2022] [Indexed: 11/24/2022] Open
Abstract
The treatment landscape of multiple myeloma (MM) has evolved considerably with the FDA-approval of at least 15 drugs over the past two decades. Together with the use of autologous stem cell transplantation, these novel therapies have resulted in significant survival benefit for patients with MM. In particular, our improved understanding of the BM and immune microenvironment has led to the development of highly effective immunotherapies that have demonstrated unprecedented response rates even in the multiple refractory disease setting. However, MM remains challenging to treat especially in a high-risk setting. A key mediator of therapeutic resistance in MM is the bone marrow (BM) microenvironment; a deeper understanding is necessary to facilitate the development of therapies that target MM in the context of the BM milieu to elicit deeper and more durable responses with the ultimate goal of long-term control or a cure of MM. In this review, we discuss our current understanding of the role the BM microenvironment plays in MM pathogenesis, with a focus on its immunosuppressive nature. We also review FDA-approved immunotherapies currently in clinical use and highlight promising immunotherapeutic approaches on the horizon.
Collapse
Affiliation(s)
- Matthew Ho
- Department of Internal Medicine, Mayo Clinic, Rochester, MN 55902, USA
| | - Alexander Xiao
- Department of Internal Medicine, Mayo Clinic, Rochester, MN 55902, USA
| | - Dongni Yi
- Department of Internal Medicine, Mayo Clinic, Rochester, MN 55902, USA
| | - Saurabh Zanwar
- Division of Hematology, Department of Internal Medicine, Mayo Clinic, Rochester, MN 55902, USA
| | - Giada Bianchi
- Division of Hematology, Brigham and Women’s Hospital, Harvard Medical School, Boston, MA 02120, USA
| |
Collapse
|
11
|
Cho SF, Yeh TJ, Anderson KC, Tai YT. Bispecific antibodies in multiple myeloma treatment: A journey in progress. Front Oncol 2022; 12:1032775. [PMID: 36330495 PMCID: PMC9623099 DOI: 10.3389/fonc.2022.1032775] [Citation(s) in RCA: 23] [Impact Index Per Article: 11.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/31/2022] [Accepted: 10/03/2022] [Indexed: 07/29/2023] Open
Abstract
The incorporation of novel agents and monoclonal antibody-based therapies into the treatment of multiple myeloma (MM) has significantly improved long-term patient survival. However, the disease is still largely incurable, with high-risk patients suffering shorter survival times, partly due to weakened immune systems. Bispecific molecules, including bispecific antibodies (BisAbs) and bispecific T-cell engagers (BiTEs), encourage immune cells to lyse MM cells by simultaneously binding antigens on MM cells and immune effector cells, bringing those cells into close proximity. BisAbs that target B-cell maturation antigen (BCMA) and GPRC5D have shown impressive clinical activity, and the results of early-phase clinical trials targeting FcRH5 in patients with relapsed/refractory MM (RRMM) are also promising. Furthermore, the safety profile of these agents is favorable, including mainly low-grade cytokine release syndrome (CRS). These off-the-shelf bispecific molecules will likely become an essential part of the MM treatment paradigm. Here, we summarize and highlight various bispecific immunotherapies under development in MM treatment, as well as the utility of combining them with current standard-of-care treatments and new strategies. With the advancement of novel combination treatment approaches, these bispecific molecules may lead the way to a cure for MM.
Collapse
Affiliation(s)
- Shih-Feng Cho
- Division of Hematology & Oncology, Department of Internal Medicine, Kaohsiung Medical University Hospital, Kaohsiung Medical University, Kaohsiung, Taiwan
- Faculty of Medicine, College of Medicine, Kaohsiung Medical University, Kaohsiung, Taiwan
- Center for Cancer Research, Kaohsiung Medical University, Kaohsiung, Taiwan
| | - Tsung-Jang Yeh
- Division of Hematology & Oncology, Department of Internal Medicine, Kaohsiung Medical University Hospital, Kaohsiung Medical University, Kaohsiung, Taiwan
| | - Kenneth C. Anderson
- LeBow Institute for Myeloma Therapeutics and Jerome Lipper Multiple Myeloma Center, Dana-Farber Cancer Institute, Harvard Medical School, Boston, MA, United States
| | - Yu-Tzu Tai
- LeBow Institute for Myeloma Therapeutics and Jerome Lipper Multiple Myeloma Center, Dana-Farber Cancer Institute, Harvard Medical School, Boston, MA, United States
| |
Collapse
|
12
|
Dima D, Jiang D, Singh DJ, Hasipek M, Shah HS, Ullah F, Khouri J, Maciejewski JP, Jha BK. Multiple Myeloma Therapy: Emerging Trends and Challenges. Cancers (Basel) 2022; 14:cancers14174082. [PMID: 36077618 PMCID: PMC9454959 DOI: 10.3390/cancers14174082] [Citation(s) in RCA: 26] [Impact Index Per Article: 13.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/15/2022] [Revised: 08/16/2022] [Accepted: 08/18/2022] [Indexed: 11/16/2022] Open
Abstract
Multiple myeloma (MM) is a complex hematologic malignancy characterized by the uncontrolled proliferation of clonal plasma cells in the bone marrow that secrete large amounts of immunoglobulins and other non-functional proteins. Despite decades of progress and several landmark therapeutic advancements, MM remains incurable in most cases. Standard of care frontline therapies have limited durable efficacy, with the majority of patients eventually relapsing, either early or later. Induced drug resistance via up-modulations of signaling cascades that circumvent the effect of drugs and the emergence of genetically heterogeneous sub-clones are the major causes of the relapsed-refractory state of MM. Cytopenias from cumulative treatment toxicity and disease refractoriness limit therapeutic options, hence creating an urgent need for innovative approaches effective against highly heterogeneous myeloma cell populations. Here, we present a comprehensive overview of the current and future treatment paradigm of MM, and highlight the gaps in therapeutic translations of recent advances in targeted therapy and immunotherapy. We also discuss the therapeutic potential of emerging preclinical research in multiple myeloma.
Collapse
Affiliation(s)
- Danai Dima
- Department of Translational Hematology and Oncology Research, Taussig Cancer Institute, Cleveland Clinic, Cleveland, OH 44195, USA
- Center for Immunotherapy and Precision Immuno-Oncology, Lerner Research Institute, Cleveland, OH 44195, USA
- Department of Hematology and Medical Oncology, Taussig Cancer Institute, Cleveland Clinic, Cleveland, OH 44195, USA
| | - Dongxu Jiang
- Department of Translational Hematology and Oncology Research, Taussig Cancer Institute, Cleveland Clinic, Cleveland, OH 44195, USA
- Center for Immunotherapy and Precision Immuno-Oncology, Lerner Research Institute, Cleveland, OH 44195, USA
| | - Divya Jyoti Singh
- Department of Translational Hematology and Oncology Research, Taussig Cancer Institute, Cleveland Clinic, Cleveland, OH 44195, USA
- Center for Immunotherapy and Precision Immuno-Oncology, Lerner Research Institute, Cleveland, OH 44195, USA
| | - Metis Hasipek
- Department of Translational Hematology and Oncology Research, Taussig Cancer Institute, Cleveland Clinic, Cleveland, OH 44195, USA
| | - Haikoo S. Shah
- Department of Hematology and Medical Oncology, Taussig Cancer Institute, Cleveland Clinic, Cleveland, OH 44195, USA
| | - Fauzia Ullah
- Department of Translational Hematology and Oncology Research, Taussig Cancer Institute, Cleveland Clinic, Cleveland, OH 44195, USA
| | - Jack Khouri
- Department of Hematology and Medical Oncology, Taussig Cancer Institute, Cleveland Clinic, Cleveland, OH 44195, USA
- Case Comprehensive Cancer Center, Case Western Reserve University, Cleveland, OH 44106, USA
- Cleveland Clinic Lerner College of Medicine, Cleveland, OH 44195, USA
| | - Jaroslaw P. Maciejewski
- Department of Translational Hematology and Oncology Research, Taussig Cancer Institute, Cleveland Clinic, Cleveland, OH 44195, USA
- Case Comprehensive Cancer Center, Case Western Reserve University, Cleveland, OH 44106, USA
- Cleveland Clinic Lerner College of Medicine, Cleveland, OH 44195, USA
| | - Babal K. Jha
- Department of Translational Hematology and Oncology Research, Taussig Cancer Institute, Cleveland Clinic, Cleveland, OH 44195, USA
- Center for Immunotherapy and Precision Immuno-Oncology, Lerner Research Institute, Cleveland, OH 44195, USA
- Case Comprehensive Cancer Center, Case Western Reserve University, Cleveland, OH 44106, USA
- Cleveland Clinic Lerner College of Medicine, Cleveland, OH 44195, USA
- Correspondence:
| |
Collapse
|
13
|
T-cell redirecting bispecific antibodies in multiple myeloma: a revolution? Blood 2022; 139:3681-3687. [PMID: 35404996 DOI: 10.1182/blood.2021014611] [Citation(s) in RCA: 13] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/24/2022] [Accepted: 03/01/2022] [Indexed: 11/20/2022] Open
Abstract
Bispecific antibodies are designed to link a surface target molecule on the malignant plasma cells to CD3 on T-cells and thereby redirect activated T-cells to induce tumor cell death. Early-phase clinical trials targeting B-cell maturation antigen, GPRC5D or FcRH5, have demonstrated a favorable safety profile and promising efficacy data in triple-class refractory multiple myeloma. This novel immunotherapeutic modality will likely change the treatment paradigm in the coming years.
Collapse
|
14
|
Natale V, Stadlmayr G, Benedetti F, Stadlbauer K, Rüker F, Wozniak-Knopp G. Trispecific antibodies produced from mAb 2 pairs by controlled Fab-arm exchange. Biol Chem 2022; 403:509-523. [PMID: 35089662 DOI: 10.1515/hsz-2021-0376] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/28/2021] [Accepted: 01/18/2022] [Indexed: 12/13/2022]
Abstract
Bispecific antibodies and antibody fragments are therapeutics of growing importance. They are clinically applied for effector cell engagement, enhanced targeting selectivity, addressing of multiple cellular pathways and active transfer of certain activities into difficult-to-reach compartments. These functionalities could profit from a third antigen specificity. In this work we have employed symmetrical bispecific parental antibodies of mAb2 format, which feature a novel antigen binding site in the CH3 domains, and engineered them with a minimal number of point mutations to guide the formation of a controlled Fab-arm exchanged trispecific antibody at a high yield after reduction and re-oxidation. Two model antibodies, one reactive with EGFR, Her2 and VEGF, and one with Fab-arms binding to Ang2 and VEGF and an Fc fragment binding to VEGF, were prepared and examined for heterodimeric status, stability, antigen binding properties and biological activity. Resulting molecules were of good biophysical characteristics and retained antigen reactivity and biological activity of the parental mAb2 constructs.
Collapse
Affiliation(s)
- Veronica Natale
- Department of Biotechnology, Christian Doppler Laboratory for Innovative Immunotherapeutics, Institute of Molecular Biotechnology, University of Natural Resources and Life Sciences, Vienna (BOKU), Muthgasse 18, A-1190 Vienna, Austria
| | - Gerhard Stadlmayr
- Department of Biotechnology, Christian Doppler Laboratory for Innovative Immunotherapeutics, Institute of Molecular Biotechnology, University of Natural Resources and Life Sciences, Vienna (BOKU), Muthgasse 18, A-1190 Vienna, Austria
| | - Filippo Benedetti
- Department of Biotechnology, Christian Doppler Laboratory for Innovative Immunotherapeutics, Institute of Molecular Biotechnology, University of Natural Resources and Life Sciences, Vienna (BOKU), Muthgasse 18, A-1190 Vienna, Austria
| | - Katharina Stadlbauer
- Department of Biotechnology, Christian Doppler Laboratory for Innovative Immunotherapeutics, Institute of Molecular Biotechnology, University of Natural Resources and Life Sciences, Vienna (BOKU), Muthgasse 18, A-1190 Vienna, Austria
| | - Florian Rüker
- Department of Biotechnology, Christian Doppler Laboratory for Innovative Immunotherapeutics, Institute of Molecular Biotechnology, University of Natural Resources and Life Sciences, Vienna (BOKU), Muthgasse 18, A-1190 Vienna, Austria
| | - Gordana Wozniak-Knopp
- Department of Biotechnology, Christian Doppler Laboratory for Innovative Immunotherapeutics, Institute of Molecular Biotechnology, University of Natural Resources and Life Sciences, Vienna (BOKU), Muthgasse 18, A-1190 Vienna, Austria
| |
Collapse
|
15
|
Abstract
Multiple myeloma is the second most common hematological malignancy in adults, accounting for 2% of all cancer-related deaths in the UK. Current chemotherapy-based regimes are insufficient, as most patients relapse and develop therapy resistance. This review focuses on current novel antibody- and aptamer-based therapies aiming to overcome current therapy limitations, as well as their respective limitations and areas of improvement. The use of computer modeling methods, as a tool to study and improve ligand-receptor alignments for the use of novel therapy development will also be discussed, as it has become a rapid, reliable and comparatively inexpensive method of investigation.
Collapse
|
16
|
Krejcik J, Barnkob MB, Nyvold CG, Larsen TS, Barington T, Abildgaard N. Harnessing the Immune System to Fight Multiple Myeloma. Cancers (Basel) 2021; 13:4546. [PMID: 34572773 PMCID: PMC8467095 DOI: 10.3390/cancers13184546] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/21/2021] [Revised: 09/03/2021] [Accepted: 09/06/2021] [Indexed: 12/14/2022] Open
Abstract
Multiple myeloma (MM) is a heterogeneous plasma cell malignancy differing substantially in clinical behavior, prognosis, and response to treatment. With the advent of novel therapies, many patients achieve long-lasting remissions, but some experience aggressive and treatment refractory relapses. So far, MM is considered incurable. Myeloma pathogenesis can broadly be explained by two interacting mechanisms, intraclonal evolution of cancer cells and development of an immunosuppressive tumor microenvironment. Failures in isotype class switching and somatic hypermutations result in the neoplastic transformation typical of MM and other B cell malignancies. Interestingly, although genetic alterations occur and evolve over time, they are also present in premalignant stages, which never progress to MM, suggesting that genetic mutations are necessary but not sufficient for myeloma transformation. Changes in composition and function of the immune cells are associated with loss of effective immune surveillance, which might represent another mechanism driving malignant transformation. During the last decade, the traditional view on myeloma treatment has changed dramatically. It is increasingly evident that treatment strategies solely based on targeting intrinsic properties of myeloma cells are insufficient. Lately, approaches that redirect the cells of the otherwise suppressed immune system to take control over myeloma have emerged. Evidence of utility of this principle was initially established by the observation of the graft-versus-myeloma effect in allogeneic stem cell-transplanted patients. A variety of new strategies to harness both innate and antigen-specific immunity against MM have recently been developed and intensively tested in clinical trials. This review aims to give readers a basic understanding of how the immune system can be engaged to treat MM, to summarize the main immunotherapeutic modalities, their current role in clinical care, and future prospects.
Collapse
Affiliation(s)
- Jakub Krejcik
- Centre for Cellular Immunotherapy of Haematological Cancer Odense (CITCO), Odense University Hospital, 5000 Odense, Denmark; (J.K.); (M.B.B.); (C.G.N.); (T.S.L.); (T.B.)
- Department of Haematology, Odense University Hospital, 5000 Odense, Denmark
- Haematology Research Unit, Department of Clinical Research, University of Southern Denmark, 5000 Odense, Denmark
| | - Mike Bogetofte Barnkob
- Centre for Cellular Immunotherapy of Haematological Cancer Odense (CITCO), Odense University Hospital, 5000 Odense, Denmark; (J.K.); (M.B.B.); (C.G.N.); (T.S.L.); (T.B.)
- Department of Clinical Immunology, Odense University Hospital, 5000 Odense, Denmark
| | - Charlotte Guldborg Nyvold
- Centre for Cellular Immunotherapy of Haematological Cancer Odense (CITCO), Odense University Hospital, 5000 Odense, Denmark; (J.K.); (M.B.B.); (C.G.N.); (T.S.L.); (T.B.)
- Haematology Research Unit, Department of Clinical Research, University of Southern Denmark, 5000 Odense, Denmark
- Haematology-Pathology Research Laboratory, Research Unit for Haematology and Research Unit for Pathology, University of Southern Denmark and Odense University Hospital, 5000 Odense, Denmark
| | - Thomas Stauffer Larsen
- Centre for Cellular Immunotherapy of Haematological Cancer Odense (CITCO), Odense University Hospital, 5000 Odense, Denmark; (J.K.); (M.B.B.); (C.G.N.); (T.S.L.); (T.B.)
- Department of Haematology, Odense University Hospital, 5000 Odense, Denmark
- Haematology Research Unit, Department of Clinical Research, University of Southern Denmark, 5000 Odense, Denmark
| | - Torben Barington
- Centre for Cellular Immunotherapy of Haematological Cancer Odense (CITCO), Odense University Hospital, 5000 Odense, Denmark; (J.K.); (M.B.B.); (C.G.N.); (T.S.L.); (T.B.)
- Department of Clinical Immunology, Odense University Hospital, 5000 Odense, Denmark
| | - Niels Abildgaard
- Centre for Cellular Immunotherapy of Haematological Cancer Odense (CITCO), Odense University Hospital, 5000 Odense, Denmark; (J.K.); (M.B.B.); (C.G.N.); (T.S.L.); (T.B.)
- Department of Haematology, Odense University Hospital, 5000 Odense, Denmark
- Haematology Research Unit, Department of Clinical Research, University of Southern Denmark, 5000 Odense, Denmark
| |
Collapse
|
17
|
Abramson HN. Immunotherapy of Multiple Myeloma: Promise and Challenges. Immunotargets Ther 2021; 10:343-371. [PMID: 34527606 PMCID: PMC8437262 DOI: 10.2147/itt.s306103] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/15/2021] [Accepted: 08/25/2021] [Indexed: 12/16/2022] Open
Abstract
Whereas the treatment of MM was dependent solely on alkylating agents and corticosteroids during the prior three decades, the landscape of therapeutic measures to treat the disease began to expand enormously early in the current century. The introduction of new classes of small-molecule drugs, such as proteasome blockers (bortezomib and carfilzomib), immunomodulators (lenalidomide and pomalidomide), nuclear export inhibitors (selinexor), and histone deacetylase blockers (panobinostat), as well as the application of autologous stem cell transplantation (ASCT), resulted in a seismic shift in how the disease is treated. The picture changed dramatically once again starting with the 2015 FDA approval of two monoclonal antibodies (mAbs) - the anti-CD38 daratumumab and the anti-SLAMF7 elotuzumab. Daratumumab, in particular, has had a great impact on MM therapy and today is often included in various regimens to treat the disease, both in newly diagnosed cases and in the relapse/refractory setting. Recently, other immunotherapies have been added to the arsenal of drugs available to fight this malignancy. These include isatuximab (also anti-CD38) and, in the past year, the antibody-drug conjugate (ADC) belantamab mafodotin and the chimeric antigen receptor (CAR) T-cell product idecabtagene vicleucel (ide-cel). While the accumulated benefits of these newer agents have resulted in a doubling of the disease's five-year survival rate to more than 5 years and improved quality of life, the disease remains incurable. Almost without exception patients experience relapse and/or become refractory to the drugs used, making the search for innovative therapies all the more essential. This review covers the current scope of anti-myeloma immunotherapeutic agents, both those in clinical use and on the horizon, including naked mAbs, ADCs, bi- and multi-targeted mAbs, and CAR T-cells. Emphasis is placed on the benefits of each along with the challenges that need to be overcome if MM is to be considered curable in the future.
Collapse
Affiliation(s)
- Hanley N Abramson
- Wayne State University, Department of Pharmaceutical Sciences, Detroit, MI, 48201, USA
| |
Collapse
|
18
|
Lancman G, Sastow DL, Cho HJ, Jagannath S, Madduri D, Parekh SS, Richard S, Richter J, Sanchez L, Chari A. Bispecific Antibodies in Multiple Myeloma: Present and Future. Blood Cancer Discov 2021; 2:423-433. [PMID: 34661161 PMCID: PMC8510808 DOI: 10.1158/2643-3230.bcd-21-0028] [Citation(s) in RCA: 53] [Impact Index Per Article: 17.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/23/2021] [Revised: 05/06/2021] [Accepted: 07/07/2021] [Indexed: 11/16/2022] Open
Abstract
Despite many recent advances in therapy, there is still no plateau in overall survival curves in multiple myeloma. Bispecific antibodies are a novel immunotherapeutic approach designed to bind antigens on malignant plasma cells and cytotoxic immune effector cells. Early-phase clinical trials targeting B-cell maturation antigen (BCMA), GPRC5D, and FcRH5 have demonstrated a favorable safety profile, with mainly low-grade cytokine release syndrome, cytopenias, and infections. Although dose escalation is ongoing in several studies, early efficacy data show response rates in the most active dose cohorts between 61% and 83% with many deep responses; however, durability remains to be established. Further clinical trial data are eagerly anticipated. SIGNIFICANCE Overall survival of triple-class refractory multiple myeloma remains poor. Bispecific antibodies are a novel immunotherapeutic modality with a favorable safety profile and impressive preliminary efficacy in heavily treated patients. Although more data are needed, bispecifics will likely become an integral part of the multiple myeloma treatment paradigm in the near future. Studies in earlier lines of therapy and in combination with other active anti-multiple myeloma agents will help further define the role of bispecifics in multiple myeloma.
Collapse
Affiliation(s)
- Guido Lancman
- Tisch Cancer Insitute, Icahn School of Medicine at Mount Sinai, New York, New York
| | | | - Hearn J Cho
- Tisch Cancer Insitute, Icahn School of Medicine at Mount Sinai, New York, New York
| | - Sundar Jagannath
- Tisch Cancer Insitute, Icahn School of Medicine at Mount Sinai, New York, New York
| | - Deepu Madduri
- Tisch Cancer Insitute, Icahn School of Medicine at Mount Sinai, New York, New York
| | - Samir S Parekh
- Tisch Cancer Insitute, Icahn School of Medicine at Mount Sinai, New York, New York
| | - Shambavi Richard
- Tisch Cancer Insitute, Icahn School of Medicine at Mount Sinai, New York, New York
| | - Joshua Richter
- Tisch Cancer Insitute, Icahn School of Medicine at Mount Sinai, New York, New York
| | - Larysa Sanchez
- Tisch Cancer Insitute, Icahn School of Medicine at Mount Sinai, New York, New York
| | - Ajai Chari
- Tisch Cancer Insitute, Icahn School of Medicine at Mount Sinai, New York, New York.
| |
Collapse
|
19
|
Wingert S, Reusch U, Knackmuss S, Kluge M, Damrat M, Pahl J, Schniegler-Mattox U, Mueller T, Fucek I, Ellwanger K, Tesar M, Haneke T, Koch J, Treder M, Fischer W, Rajkovic E. Preclinical evaluation of AFM24, a novel CD16A-specific innate immune cell engager targeting EGFR-positive tumors. MAbs 2021; 13:1950264. [PMID: 34325617 PMCID: PMC8331026 DOI: 10.1080/19420862.2021.1950264] [Citation(s) in RCA: 35] [Impact Index Per Article: 11.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022] Open
Abstract
Epidermal growth factor receptor (EGFR)-targeted cancer therapy such as anti-EGFR monoclonal antibodies and tyrosine kinase inhibitors have demonstrated clinical efficacy. However, there remains a medical need addressing limitations of these therapies, which include a narrow therapeutic window mainly due to skin and organ toxicity, and primary and secondary resistance mechanisms of the EGFR-signaling cascade (e.g., RAS-mutated colorectal cancer). Using the redirected optimized cell killing (ROCK®) antibody platform, we have developed AFM24, a novel bispecific, IgG1-scFv fusion antibody targeting CD16A on innate immune cells, and EGFR on tumor cells. We herein demonstrate binding of AFM24 to CD16A on natural killer (NK) cells and macrophages with KD values in the low nanomolar range and to various EGFR-expressing tumor cells. AFM24 was highly potent and effective for antibody-dependent cell-mediated cytotoxicity via NK cells, and also mediated antibody-dependent cellular phagocytosis via macrophages in vitro. Importantly, AFM24 was effective toward a variety of EGFR-expressing tumor cells, regardless of EGFR expression level and KRAS/BRAF mutational status. In vivo, AFM24 was well tolerated up to the highest dose (75 mg/kg) when administered to cynomolgus monkeys once weekly for 28 days. Notably, skin and other toxicities were not observed. A transient elevation of interleukin-6 levels was detected at all dose levels, 2-4 hours post-dose, which returned to baseline levels after 24 hours. These results emphasize the promise of bispecific innate cell engagers as an alternative cancer therapy and demonstrate the potential for AFM24 to effectively target tumors expressing varying levels of EGFR, regardless of their mutational status.Abbreviations: ADA: antidrug antibody; ADCC: antibody-dependent cell-mediated cytotoxicity; ADCP: antibody-dependent cellular phagocytosis; AUC: area under the curve; CAR: chimeric-antigen receptor; CD: Cluster of differentiation; CRC :colorectal cancer; ECD: extracellular domain; EGF: epidermal growth factorEGFR epidermal growth factor receptor; ELISA: enzyme-linked immunosorbent assay; FACS: fluorescence-activated cell sorting; Fc: fragment, crystallizableFv variable fragment; HNSCC: head and neck squamous carcinomaIL interleukinm; Ab monoclonal antibody; MOA: mechanism of action; NK :natural killer; NSCLC: non-small cell lung cancer; PBMC: peripheral blood mononuclear cell; PBS: phosphate-buffered saline; PD: pharmacodynamic; ROCK: redirected optimized cell killing; RSV: respiratory syncytial virus; SABC: specific antibody binding capacity; SD: standard deviation; TAM: tumor-associated macrophage; TKI: tyrosine kinase inhibitor; WT: wildtype.
Collapse
Affiliation(s)
| | - Uwe Reusch
- Research & Development, Affimed GmbH, Heidelberg, Germany
| | | | - Michael Kluge
- Research & Development, Affimed GmbH, Heidelberg, Germany
| | - Michael Damrat
- Research & Development, Affimed GmbH, Heidelberg, Germany
| | - Jens Pahl
- Research & Development, Affimed GmbH, Heidelberg, Germany
| | | | - Thomas Mueller
- Research & Development, Affimed GmbH, Heidelberg, Germany
| | - Ivica Fucek
- Research & Development, Affimed GmbH, Heidelberg, Germany
| | | | - Michael Tesar
- Research & Development, Affimed GmbH, Heidelberg, Germany
| | - Torsten Haneke
- Research & Development, Affimed GmbH, Heidelberg, Germany
| | - Joachim Koch
- Research & Development, Affimed GmbH, Heidelberg, Germany
| | - Martin Treder
- Formerly Affimed GmbH, Heidelberg, Germany. Now: Arjuna Therapeutics, Santiago De Compostela, Spain
| | | | - Erich Rajkovic
- Research & Development, Affimed GmbH, Heidelberg, Germany
| |
Collapse
|
20
|
You G, Won J, Lee Y, Moon D, Park Y, Lee SH, Lee SW. Bispecific Antibodies: A Smart Arsenal for Cancer Immunotherapies. Vaccines (Basel) 2021; 9:724. [PMID: 34358141 PMCID: PMC8310217 DOI: 10.3390/vaccines9070724] [Citation(s) in RCA: 24] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/30/2021] [Revised: 06/05/2021] [Accepted: 06/28/2021] [Indexed: 12/13/2022] Open
Abstract
Following the clinical success of cancer immunotherapies such as immune checkpoint inhibitors blocking B7/CTLA-4 or PD-1/PD-L1 signaling and ongoing numerous combination therapies in the clinic,3 bispecific antibodies (BsAbs) are now emerging as a growing class of immunotherapies with the potential to improve clinical efficacy and safety further. Here, we describe four classes of BsAbs: (a) immune effector cell redirectors; (b) tumor-targeted immunomodulators; (c) dual immunomodulators; and (d) dual tumor-targeting BsAbs. This review describes each of these classes of BsAbs and presents examples of BsAbs in development. We reviewed the biological rationales and characteristics of BsAbs and summarized the current status and limitations of clinical development of BsAbs and strategies to overcome limitations. The field of BsAb-based cancer immunotherapy is growing, and more data from clinical trials are accumulating. Thus, BsAbs could be the next generation of new treatment options for cancer patients.
Collapse
Affiliation(s)
- Gihoon You
- Department of Life Sciences, Pohang University of Science and Technology (POSTECH), Pohang 37673, Korea; (G.Y.); (D.M.)
| | - Jonghwa Won
- ABL Bio Inc., Seongnam 13488, Korea; (J.W.); (Y.L.); (S.H.L.)
| | - Yangsoon Lee
- ABL Bio Inc., Seongnam 13488, Korea; (J.W.); (Y.L.); (S.H.L.)
| | - Dain Moon
- Department of Life Sciences, Pohang University of Science and Technology (POSTECH), Pohang 37673, Korea; (G.Y.); (D.M.)
| | - Yunji Park
- Biotechcenter, POSTECH, Pohang 37673, Korea;
| | - Sang Hoon Lee
- ABL Bio Inc., Seongnam 13488, Korea; (J.W.); (Y.L.); (S.H.L.)
| | - Seung-Woo Lee
- Department of Life Sciences, Pohang University of Science and Technology (POSTECH), Pohang 37673, Korea; (G.Y.); (D.M.)
| |
Collapse
|
21
|
Brouillard A, Deshpande N, Kulkarni AA. Engineered Multifunctional Nano- and Biological Materials for Cancer Immunotherapy. Adv Healthc Mater 2021; 10:e2001680. [PMID: 33448159 DOI: 10.1002/adhm.202001680] [Citation(s) in RCA: 12] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/21/2020] [Revised: 12/21/2020] [Indexed: 12/19/2022]
Abstract
Cancer immunotherapy is set to emerge as the future of cancer therapy. However, recent immunotherapy trials in different cancers have yielded sub-optimal results, with durable responses seen in only a small fraction of patients. Engineered multifunctional nanomaterials and biological materials are versatile platforms that can elicit strong immune responses and improve anti-cancer efficacy when applied to cancer immunotherapy. While there are traditional systems such as polymer- and lipid-based nanoparticles, there is a wide variety of other materials with inherent and additive properties that can allow for more potent activation of the immune system. By synthesizing and applying multifunctional strategies, it allows for a more extensive and more effective repertoire of tools to use in the wide variety of situations that cancer presents itself. Here, several types of nanoscale and biological material strategies and platforms that provide their inherent benefits for targeting and activating multiple aspects of the immune system are discussed. Overall, this review aims to provide a comprehensive understanding of recent advances in the field of multifunctional cancer immunotherapy and trends that pave the way for more diverse and tactical regression of tumors through soliciting responses by either the adaptive or innate immune system, and even both simultaneously.
Collapse
Affiliation(s)
- Anthony Brouillard
- Department of Chemical Engineering University of Massachusetts Amherst MA 01003 USA
| | - Nilesh Deshpande
- Department of Chemical Engineering University of Massachusetts Amherst MA 01003 USA
| | - Ashish A. Kulkarni
- Department of Chemical Engineering University of Massachusetts Amherst MA 01003 USA
- Center for Bioactive Delivery Institute for Applied Life Sciences University of Massachusetts Amherst MA 01003 USA
| |
Collapse
|
22
|
Yang Y, Li Y, Gu H, Dong M, Cai Z. Emerging agents and regimens for multiple myeloma. J Hematol Oncol 2020; 13:150. [PMID: 33168044 PMCID: PMC7654052 DOI: 10.1186/s13045-020-00980-5] [Citation(s) in RCA: 46] [Impact Index Per Article: 11.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/01/2020] [Accepted: 10/19/2020] [Indexed: 02/08/2023] Open
Abstract
The outcomes of multiple myeloma (MM) have been improved significantly with the therapies incorporating proteasome inhibitors (PI), immunomodulatory drugs, monoclonal antibodies (MoAb) and stem cell transplantation. However, relapsed and refractory MM (RRMM) remains a major challenge. Novel agents and regimens are under active clinical development. These include new PIs such as ixazomib, marizomib, and oprozomib; new MoAbs such as isatuximab and MOR202; novel epigenetic agent ricolinostat and novel cytokines such as siltuximab. Recently, the first XPO-1 inhibitor, selinexor, was approved for RRMM. BCMA-targeted BiTE, antibody-drug conjugates and CAR-T cells have the potential to revolutionize the therapy for RRMM. In this review, we summarized the latest clinical development of these novel agents and regimens.
Collapse
Affiliation(s)
- Yang Yang
- Bone Marrow Transplantation Center, Department of Hematology, The First Affiliated Hospital, College of Medicine, Zhejiang University, Hangzhou, Zhejiang, China
| | - Yi Li
- Bone Marrow Transplantation Center, Department of Hematology, The First Affiliated Hospital, College of Medicine, Zhejiang University, Hangzhou, Zhejiang, China
| | - Huiyao Gu
- Bone Marrow Transplantation Center, Department of Hematology, The First Affiliated Hospital, College of Medicine, Zhejiang University, Hangzhou, Zhejiang, China
| | - Mengmeng Dong
- Bone Marrow Transplantation Center, Department of Hematology, The First Affiliated Hospital, College of Medicine, Zhejiang University, Hangzhou, Zhejiang, China
| | - Zhen Cai
- Bone Marrow Transplantation Center, Department of Hematology, The First Affiliated Hospital, College of Medicine, Zhejiang University, Hangzhou, Zhejiang, China.
- Institute of Hematology, Zhejiang University, Hangzhou, Zhejiang, China.
- Zhejiang Laboratory for Systems and Precision Medicine, Zhejiang University Medical Center, Hangzhou, Zhejiang, China.
| |
Collapse
|
23
|
Lamendour L, Deluce-Kakwata-Nkor N, Mouline C, Gouilleux-Gruart V, Velge-Roussel F. Tethering Innate Surface Receptors on Dendritic Cells: A New Avenue for Immune Tolerance Induction? Int J Mol Sci 2020; 21:E5259. [PMID: 32722168 PMCID: PMC7432195 DOI: 10.3390/ijms21155259] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/08/2020] [Revised: 07/22/2020] [Accepted: 07/23/2020] [Indexed: 12/26/2022] Open
Abstract
Dendritic cells (DCs) play a key role in immunity and are highly potent at presenting antigens and orienting the immune response. Depending on the environmental signals, DCs could turn the immune response toward immunity or immune tolerance. Several subsets of DCs have been described, with each expressing various surface receptors and all participating in DC-associated immune functions according to their specific skills. DC subsets could also contribute to the vicious circle of inflammation in immune diseases and establishment of immune tolerance in cancer. They appear to be appropriate targets in the control of inflammatory diseases or regulation of autoimmune responses. For all these reasons, in situ DC targeting with therapeutic antibodies seems to be a suitable way of modulating the entire immune system. At present, the field of antibody-based therapies has mainly been developed in oncology, but it is undergoing remarkable expansion thanks to a wide variety of antibody formats and their related functions. Moreover, current knowledge of DC biology may open new avenues for targeting and modulating the different DC subsets. Based on an update of pathogen recognition receptor expression profiles in human DC subsets, this review evaluates the possibility of inducing tolerant DCs using antibody-based therapeutic agents.
Collapse
Affiliation(s)
| | | | | | | | - Florence Velge-Roussel
- GICC EA 7501, Université de Tours, UFR de Médecine, 10 Boulevard Tonnellé, F-37032 Tours, France; (L.L.); (N.D.-K.-N.); (C.M.); (V.G.-G.)
| |
Collapse
|
24
|
Abramson HN. B-Cell Maturation Antigen (BCMA) as a Target for New Drug Development in Relapsed and/or Refractory Multiple Myeloma. Int J Mol Sci 2020; 21:E5192. [PMID: 32707894 PMCID: PMC7432930 DOI: 10.3390/ijms21155192] [Citation(s) in RCA: 20] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/02/2020] [Revised: 07/19/2020] [Accepted: 07/20/2020] [Indexed: 02/07/2023] Open
Abstract
During the past two decades there has been a major shift in the choice of agents to treat multiple myeloma, whether newly diagnosed or in the relapsed/refractory stage. The introduction of new drug classes, such as proteasome inhibitors, immunomodulators, and anti-CD38 and anti-SLAMF7 monoclonal antibodies, coupled with autologous stem cell transplantation, has approximately doubled the disease's five-year survival rate. However, this positive news is tempered by the realization that these measures are not curative and patients eventually relapse and/or become resistant to the drug's effects. Thus, there is a need to discover newer myeloma-driving molecular markers and develop innovative drugs designed to precisely regulate the actions of such putative targets. B cell maturation antigen (BCMA), which is found almost exclusively on the surfaces of malignant plasma cells to the exclusion of other cell types, including their normal counterparts, has emerged as a specific target of interest in this regard. Immunotherapeutic agents have been at the forefront of research designed to block BCMA activity. These agents encompass monoclonal antibodies, such as the drug conjugate belantamab mafodotin; bispecific T-cell engager strategies exemplified by AMG 420; and chimeric antigen receptor (CAR) T-cell therapeutics that include idecabtagene vicleucel (bb2121) and JNJ-68284528.
Collapse
Affiliation(s)
- Hanley N Abramson
- Department of Pharmaceutical Sciences, Wayne State University, Detroit, MI 48202, USA
| |
Collapse
|
25
|
Zhou X, Einsele H, Danhof S. Bispecific Antibodies: A New Era of Treatment for Multiple Myeloma. J Clin Med 2020; 9:jcm9072166. [PMID: 32659909 PMCID: PMC7408718 DOI: 10.3390/jcm9072166] [Citation(s) in RCA: 22] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/04/2020] [Revised: 07/02/2020] [Accepted: 07/06/2020] [Indexed: 02/06/2023] Open
Abstract
Despite the introduction of novel agents such as proteasome inhibitors, immunomodulatory drugs, and autologous stem cell transplant, multiple myeloma (MM) largely remains an incurable disease. In recent years, monoclonal antibody-based treatment strategies have been developed to target specific surface antigens on MM cells. Treatment with bispecific antibodies (bsAbs) is an immunotherapeutic strategy that leads to an enhanced interaction between MM cells and immune effector cells, e.g., T-cells and natural killer cells. With the immune synapse built by bsAbs, the elimination of MM cells can be facilitated. To date, bsAbs have demonstrated encouraging results in preclinical studies, and clinical trials evaluating bsAbs in patients with MM are ongoing. Early clinical data show the promising efficacy of bsAbs in relapsed/refractory MM. Together with chimeric antigen receptor-modified (CAR)-T-cells, bsAbs represent a new dimension of precision medicine. In this review, we provide an overview of rationale, current clinical development, resistance mechanisms, and future directions of bsAbs in MM.
Collapse
|
26
|
Improving Immunotherapy Against B-Cell Malignancies Using γδ T-Cell-specific Stimulation and Therapeutic Monoclonal Antibodies. J Immunother 2020; 42:331-344. [PMID: 31318724 DOI: 10.1097/cji.0000000000000289] [Citation(s) in RCA: 16] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022]
Abstract
Tumor antigen-targeting monoclonal antibodies (mAbs) are an important element of current cancer therapies. Some of these therapeutic mAbs enable antibody-dependent cell mediated cytotoxicity (ADCC) against tumor cells. However, cancer-related functional impairment of immune effector cells may limit the clinical efficacy of antibody treatments. We reckoned that combining mAbs with cell-based immunotherapies would provide a clinically relevant synergism and benefit for cancer patients. Here, we focus on γδ T cells, as earlier studies demonstrated that γδ T-cell-based therapies are safe and promising for several types of malignancies. Similar to natural killer cells, their antitumor effects can be enhanced using antibodies, and they could, therefore, become a versatile effector cell platform for use with a variety of licensed therapeutic mAbs against cancer. In this study, we explore the potential of a combination therapy of activated γδ T cells with rituximab and the more recently developed mAbs (obinutuzumab and daratumumab) in different B-cell malignancies in vitro. Obinutuzumab outperformed the other mAbs with regard to direct target cell lysis and ADCC by γδ T cells in several CD20 cell lines and primary lymphoma specimens. We demonstrate that comparatively few CD16 γδ T cells are sufficient to mediate a strong ADCC. Using Fc-receptor-positive B-cell lymphomas as target cells, ADCC cannot be blocked by high concentrations of immunoglobulins or anti-CD16 antibodies, but both substances can promote cell mediated target cell lysis. This study expands on earlier reports on the therapeutic potential of distinctive tumor antigen-targeting mAbs and facilitates the understanding of the mechanism and potential of ADCC by γδ T-cell subsets.
Collapse
|
27
|
Abstract
Bispecific therapeutics target two distinct antigens simultaneously and provide novel functionalities that are not attainable with single monospecific molecules or combinations of them. The unique potential of bispecific therapeutics is driving extensive efforts to discover synergistic dual targets, design molecular formats to integrate bispecific elements, and accelerate successful clinical translation. In particular, the past decade has witnessed a boom in the design and development of bispecific antibody formats with more than 100 collections to date. Despite the remarkable progress that has been made to expand the number of formats, qualitative fine-tuning of bispecific formats is needed to achieve optimal dual-target engagement based on understanding of the spatiotemporal interdependence of the two physically linked binding specificities and the complex target biology associated with bispecific approaches. This review provides insights into the design parameters - including affinity, valency, and geometry - that need to be considered at an early stage of development in order to take the best advantage of bispecific therapeutics.
Collapse
Affiliation(s)
- Sung In Lim
- Department of Chemical Engineering, Pukyong National University, Yongso-ro 45, Nam-gu, Busan, South Korea.
| |
Collapse
|
28
|
Abstract
The therapeutic landscape of multiple myeloma (MM) has dramatically changed in the last 15 years with the advent of immunomodulatory drugs and proteasome inhibitors. However, majority of MM patients relapse, and new therapies are needed. Various agents with diverse mechanisms of action and distinct targets, including cellular therapies, monoclonal antibodies, and small molecules, are currently under investigation. In this review, we report novel drugs recently approved or under advanced investigation that will likely be incorporated in the future as new standard for MM treatment, focusing on their mechanisms of action, cellular targets, and stage of development.
Collapse
Affiliation(s)
- Raphaël Szalat
- Medical Oncology, Dana-Farber Cancer Institute, Harvard Medical School, Boston, MA
- Section of Hematology and Oncology, Boston Medical Center, Boston, USA
| | - Nikhil C. Munshi
- Medical Oncology, Dana-Farber Cancer Institute, Harvard Medical School, Boston, MA
- VA Boston Healthcare System, Boston, MA
| |
Collapse
|
29
|
Nie S, Wang Z, Moscoso-Castro M, D'Souza P, Lei C, Xu J, Gu J. Biology drives the discovery of bispecific antibodies as innovative therapeutics. Antib Ther 2020; 3:18-62. [PMID: 33928225 PMCID: PMC7990219 DOI: 10.1093/abt/tbaa003] [Citation(s) in RCA: 53] [Impact Index Per Article: 13.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/10/2019] [Revised: 02/06/2020] [Indexed: 12/17/2022] Open
Abstract
A bispecific antibody (bsAb) is able to bind two different targets or two distinct epitopes on the same target. Broadly speaking, bsAbs can include any single molecule entity containing dual specificities with at least one being antigen-binding antibody domain. Besides additive effect or synergistic effect, the most fascinating applications of bsAbs are to enable novel and often therapeutically important concepts otherwise impossible by using monoclonal antibodies alone or their combination. This so-called obligate bsAbs could open up completely new avenue for developing novel therapeutics. With evolving understanding of structural architecture of various natural or engineered antigen-binding immunoglobulin domains and the connection of different domains of an immunoglobulin molecule, and with greatly improved understanding of molecular mechanisms of many biological processes, the landscape of therapeutic bsAbs has significantly changed in recent years. As of September 2019, over 110 bsAbs are under active clinical development, and near 180 in preclinical development. In this review article, we introduce a system that classifies bsAb formats into 30 categories based on their antigen-binding domains and the presence or absence of Fc domain. We further review the biology applications of approximately 290 bsAbs currently in preclinical and clinical development, with the attempt to illustrate the principle of selecting a bispecific format to meet biology needs and selecting a bispecific molecule as a clinical development candidate by 6 critical criteria. Given the novel mechanisms of many bsAbs, the potential unknown safety risk and risk/benefit should be evaluated carefully during preclinical and clinical development stages. Nevertheless we are optimistic that next decade will witness clinical success of bsAbs or multispecific antibodies employing some novel mechanisms of action and deliver the promise as next wave of antibody-based therapeutics.
Collapse
Affiliation(s)
- Siwei Nie
- WuXi Biologics, 299 Fute Zhong Road, Waigaoqiao Free Trade Zone, Shanghai 200131, China and ,To whom correspondence should addressed. Jijie Guor Siwei Nie. or
| | - Zhuozhi Wang
- WuXi Biologics, 299 Fute Zhong Road, Waigaoqiao Free Trade Zone, Shanghai 200131, China and
| | | | - Paul D'Souza
- Clarivate Analytics, Friars House, 160 Blackfriars Road, London SE1 8EZ, UK
| | - Can Lei
- Clarivate Analytics, Friars House, 160 Blackfriars Road, London SE1 8EZ, UK
| | - Jianqing Xu
- WuXi Biologics, 299 Fute Zhong Road, Waigaoqiao Free Trade Zone, Shanghai 200131, China and
| | - Jijie Gu
- WuXi Biologics, 299 Fute Zhong Road, Waigaoqiao Free Trade Zone, Shanghai 200131, China and ,To whom correspondence should addressed. Jijie Guor Siwei Nie. or
| |
Collapse
|
30
|
Chun BM, Page DB, McArthur HL. Combination Immunotherapy Strategies in Breast Cancer. CURRENT BREAST CANCER REPORTS 2019. [DOI: 10.1007/s12609-019-00333-3] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/30/2022]
Abstract
Abstract
Purpose of Review
We summarize combination immunotherapy strategies for the treatment of breast cancer, with a focus on metastatic disease. First, a general overview of combination approaches is presented according to breast cancer subtype. Second, additional review of promising combination approaches is presented.
Recent Findings
Combination strategies utilizing chemotherapy or radiotherapy with immune checkpoint inhibition are being evaluated across multiple phase III trials. Dual immunotherapy strategies, such as dual immune checkpoint inhibition or combined co-stimulation/co-inhibition, have supportive preclinical evidence and are under early clinical investigation. Modulation of the immune microenvironment via cytokines and vaccination strategies, as well as locally focused treatments to enhance antigenic responses, are active areas of research.
Summary
Pre-clinical and translational research sheds new light on numerous ways the immune system may be modulated to fight against cancer. We describe current and emerging combination approaches which may improve patient outcomes in metastatic breast cancer.
Collapse
|
31
|
Chen Z, Yang Y, Liu LL, Lundqvist A. Strategies to Augment Natural Killer (NK) Cell Activity against Solid Tumors. Cancers (Basel) 2019; 11:cancers11071040. [PMID: 31340613 PMCID: PMC6678934 DOI: 10.3390/cancers11071040] [Citation(s) in RCA: 32] [Impact Index Per Article: 6.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/29/2019] [Revised: 07/15/2019] [Accepted: 07/18/2019] [Indexed: 02/06/2023] Open
Abstract
The immune system plays a crucial role to prevent local growth and dissemination of cancer. Therapies based on activating the immune system can result in beneficial responses in patients with metastatic disease. Treatment with antibodies targeting the immunological checkpoint axis PD-1 / PD-L1 can result in the induction of anti-tumor T cell activation leading to meaningful long-lasting clinical responses. Still, many patients acquire resistance or develop dose-limiting toxicities to these therapies. Analysis of tumors from patients who progress on anti-PD-1 treatment reveal defective interferon-signaling and antigen presentation, resulting in immune escape from T cell-mediated attack. Natural killer (NK) cells are innate lymphocytes that can kill tumor cells without prior sensitization to antigens and can be activated to kill tumor cells that have an impaired antigen processing and presentation machinery. Thus, NK cells may serve as useful effectors against tumor cells that have become resistant to classical immune checkpoint therapy. Various approaches to activate NK cells are being increasingly explored in clinical trials against cancer. While clinical benefit has been demonstrated in patients with acute myeloid leukemia receiving haploidentical NK cells, responses in patients with solid tumors are so far less encouraging. Several hurdles need to be overcome to provide meaningful clinical responses in patients with solid tumors. Here we review the recent developments to augment NK cell responses against solid tumors with regards to cytokine therapy, adoptive infusion of NK cells, NK cell engagers, and NK cell immune checkpoints.
Collapse
Affiliation(s)
- Ziqing Chen
- Department of Oncology-Pathology, Karolinska Institutet, S-17164 Stockholm, Sweden
| | - Ying Yang
- Department of Oncology-Pathology, Karolinska Institutet, S-17164 Stockholm, Sweden
| | - Lisa L Liu
- Department of Oncology-Pathology, Karolinska Institutet, S-17164 Stockholm, Sweden.
| | - Andreas Lundqvist
- Department of Oncology-Pathology, Karolinska Institutet, S-17164 Stockholm, Sweden.
| |
Collapse
|
32
|
Swan D, Lynch K, Gurney M, O’Dwyer M. Current and emerging immunotherapeutic approaches to the treatment of multiple myeloma. Ther Adv Hematol 2019; 10:2040620719854171. [PMID: 31244984 PMCID: PMC6582283 DOI: 10.1177/2040620719854171] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/11/2019] [Accepted: 05/09/2019] [Indexed: 11/17/2022] Open
Abstract
Multiple myeloma (MM) has a worldwide incidence of 1-5/100,000/year. Outcomes have improved significantly in recent years following incorporation of immunomodulatory drugs and proteasome inhibitors into standard-of-care regimes. MM is profoundly immunosuppressive, enabling immune evasion, proliferation and disease progression. The role of the immune system in MM is becoming increasingly characterized and understood, and numerous therapies are under development or in routine clinical use targeting these elements of MM pathogenesis. In this review we discuss the immunosuppressive effects of MM, then the therapies targeting these defects. Specifically, we review the monoclonal and bispecific antibodies, alongside adoptive cellular therapies currently under investigation.
Collapse
Affiliation(s)
- Dawn Swan
- Clinical Research Facility, University Hospital Galway, Newcastle Road, Galway, H91 YR71, Ireland
| | - Kevin Lynch
- National University of Ireland, Galway, Ireland
| | - Mark Gurney
- University Hospital Galway, Ireland
- National University of Ireland, Galway, Ireland
| | - Michael O’Dwyer
- University Hospital Galway, Ireland
- National University of Ireland, Galway, Ireland
| |
Collapse
|
33
|
Labrijn AF, Janmaat ML, Reichert JM, Parren PWHI. Bispecific antibodies: a mechanistic review of the pipeline. Nat Rev Drug Discov 2019; 18:585-608. [DOI: 10.1038/s41573-019-0028-1] [Citation(s) in RCA: 493] [Impact Index Per Article: 98.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/25/2022]
|
34
|
Ellwanger K, Reusch U, Fucek I, Wingert S, Ross T, Müller T, Schniegler-Mattox U, Haneke T, Rajkovic E, Koch J, Treder M, Tesar M. Redirected optimized cell killing (ROCK®): A highly versatile multispecific fit-for-purpose antibody platform for engaging innate immunity. MAbs 2019; 11:899-918. [PMID: 31172847 PMCID: PMC6601565 DOI: 10.1080/19420862.2019.1616506] [Citation(s) in RCA: 48] [Impact Index Per Article: 9.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/07/2023] Open
Abstract
Redirection of immune cells to efficiently eliminate tumor cells holds great promise. Natural killer cells (NK), macrophages, or T cells are specifically engaged with target cells expressing markers after infection or neoplastic transformation, resulting in their activation and subsequent killing of those targets. Multiple strategies to redirect immunity have been developed in the past two decades, but they have technical hurdles or cause undesirable side-effects, as exemplified by the T cell-based chimeric antigen receptor approaches (CAR-T therapies) or bispecific T cell engager platforms. Our first-in-class bispecific antibody redirecting innate immune cells to tumors (AFM13, a CD30/CD16A-specific innate immune cell engager) has shown signs of clinical efficacy in CD30-positive lymphomas and the potential to be safely administered, indicating a wider therapeutic window compared to T cell engaging therapies. AFM13 is the most advanced candidate from our fit-for-purpose redirected optimized cell killing (ROCK®) antibody platform, which comprises a plethora of CD16A-binding innate immune cell engagers with unique properties. Here, we discuss aspects of this modular platform, including the advantages of innate immune cell engagement over classical monoclonal antibodies and other engager concepts. We also present details on its potential to engineer a fit-for-purpose innate immune cell engager format that can be equipped with unique CD16A domains, modules that influence pharmacokinetic properties and molecular architectures that influence the activation of immune effectors, as well as tumor targeting. The ROCK® platform is aimed at the activation of innate immunity for the effective lysis of tumor cells and holds the promise of overcoming limitations of other approaches that redirect immune cells by widening the therapeutic window.
Collapse
Affiliation(s)
| | - Uwe Reusch
- a Affimed GmbH, Research Department , Heidelberg , Germany
| | - Ivica Fucek
- a Affimed GmbH, Research Department , Heidelberg , Germany
| | | | - Thorsten Ross
- a Affimed GmbH, Research Department , Heidelberg , Germany
| | - Thomas Müller
- a Affimed GmbH, Research Department , Heidelberg , Germany
| | | | - Torsten Haneke
- a Affimed GmbH, Research Department , Heidelberg , Germany
| | - Erich Rajkovic
- a Affimed GmbH, Research Department , Heidelberg , Germany
| | - Joachim Koch
- a Affimed GmbH, Research Department , Heidelberg , Germany
| | - Martin Treder
- a Affimed GmbH, Research Department , Heidelberg , Germany
| | - Michael Tesar
- a Affimed GmbH, Research Department , Heidelberg , Germany
| |
Collapse
|
35
|
Abstract
Bispecific antibodies have moved from being an academic curiosity with therapeutic promise to reality, with two molecules being currently commercialized (Hemlibra® and Blincyto®) and many more in clinical trials. The success of bispecific antibodies is mainly due to the continuously growing number of mechanisms of actions (MOA) they enable that are not accessible to monoclonal antibodies. One of the earliest MOA of bispecific antibodies and currently the one with the largest number of clinical trials is the redirecting of the cytotoxic activity of T-cells for oncology applications, now extending its use in infective diseases. The use of bispecific antibodies for crossing the blood-brain barrier is another important application because of its potential to advance the therapeutic options for neurological diseases. Another noteworthy application due to its growing trend is enabling a more tissue-specific delivery or activity of antibodies. The different molecular solutions to the initial hurdles that limited the development of bispecific antibodies have led to the current diverse set of bispecific or multispecific antibody formats that can be grouped into three main categories: IgG-like formats, antibody fragment-based formats, or appended IgG formats. The expanded applications of bispecific antibodies come at the price of additional challenges for clinical development. The rising complexity in their structure may increase the risk of immunogenicity and the multiple antigen specificity complicates the selection of relevant species for safety assessment.
Collapse
Affiliation(s)
- Bushra Husain
- Protein Chemistry Department, Genentech Inc., South San Francisco, CA, 94080, USA
| | - Diego Ellerman
- Protein Chemistry Department, Genentech Inc., South San Francisco, CA, 94080, USA.
| |
Collapse
|
36
|
Viardot A, Bargou R. Bispecific antibodies in haematological malignancies. Cancer Treat Rev 2018; 65:87-95. [PMID: 29635163 DOI: 10.1016/j.ctrv.2018.04.002] [Citation(s) in RCA: 49] [Impact Index Per Article: 8.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/08/2018] [Accepted: 04/02/2018] [Indexed: 12/11/2022]
Abstract
Bispecific antibodies (bsAbs) combine the binding sites of two monoclonal antibodies in one molecule. The close proximity of a tumor specific antigen and an effector cell antigen results in a targeted activation of effector cells. The mechanism is similar to the chimeric antigen receptor (CAR) T-cells, recently approved in two haematologic cancers. CAR T-cells and bsAb represent the most powerful tools for major-histocompatibility complex (MHC) independent T-cell immune response against cancer. In contrast to CAR T-cells, bsAbs are "off the shelf" drugs. As a drawback, the efficacy is dependent on a prolonged application. More than 40 years of intensive research generate a plethora of bispecific constructs with a remarkable difference in manufacturability, stability, half-life time and receptor affinity. Blinatumomab was the first approved bsAb in relapsed and refractory acute lymphoblastic leukemia. By the mature experience of blinatumomab in more than 10 clinical trials over more than one decade, we learned some lessons on how to use this new principle. The efficacy is higher in patients with less tumor burden, suggesting the use as consolidation more than for initial debulking. Main resistance mechanisms are extramedullary relapses and the expression of the inhibitory PD-L1 molecule, suggesting the value of combination with checkpoint inhibitors. CD19 loss is infrequent after blinatumomab, preserving the option for alternative CD19-direct treatments. New bsAbs in lymphoma, myeloma and acute myeloid leukemia enter phase-I trials, together with many new constructs in solid cancer.
Collapse
Affiliation(s)
- Andreas Viardot
- Department of Internal Medicine III, University Hospital Ulm, Ulm, Germany.
| | - Ralf Bargou
- Comprehensive Cancer Center Mainfranken, Universitätsklinikum Würzburg, Würzburg, Germany
| |
Collapse
|
37
|
Dahlén E, Veitonmäki N, Norlén P. Bispecific antibodies in cancer immunotherapy. Ther Adv Vaccines Immunother 2018; 6:3-17. [PMID: 29998217 DOI: 10.1177/2515135518763280] [Citation(s) in RCA: 132] [Impact Index Per Article: 22.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/09/2017] [Accepted: 02/07/2018] [Indexed: 12/29/2022] Open
Abstract
Following the clinical success of immune checkpoint antibodies targeting CTLA-4, PD-1 or PD-L1 in cancer treatment, bispecific antibodies are now emerging as a growing class of immunotherapies with potential to further improve clinical efficacy and safety. We describe three classes of immunotherapeutic bispecific antibodies: (a) cytotoxic effector cell redirectors; (b) tumor-targeted immunomodulators; and (c) dual immunomodulators. Cytotoxic effector cell redirectors are dominated by T-cell redirecting compounds, bispecific compounds engaging a tumor-associated antigen and the T-cell receptor/CD3 complex, thereby redirecting T-cell cytotoxicity to malignant cells. This is the most established class of bispecific immunotherapies, with two compounds having reached the market and numerous compounds in clinical development. Tumor-targeted immunomodulators are bispecific compounds binding to a tumor-associated antigen and an immunomodulating receptor, such as CD40 or 4-1BB. Such compounds are usually designed to be inactive until binding the tumor antigen, thereby localizing immune stimulation to the tumor environment, while minimizing immune activation elsewhere. This is expected to induce powerful activation of tumor-specific T cells with reduced risk of immune-related adverse events. Finally, dual immunomodulators are bispecific compounds that bind two distinct immunomodulating targets, often combining targeting of PD-1 or PD-L1 with that of LAG-3 or TIM-3. The rationale is to induce superior tumor immunity compared to monospecific antibodies to the same targets. In this review, we describe each of these classes of bispecific antibodies, and present examples of compounds in development.
Collapse
Affiliation(s)
- Eva Dahlén
- Alligator Bioscience, 22381 Lund, Sweden
| | | | - Per Norlén
- Alligator Bioscience, 22381 Lund, Sweden
| |
Collapse
|
38
|
Tumor target amplification: Implications for nano drug delivery systems. J Control Release 2018; 275:142-161. [PMID: 29454742 DOI: 10.1016/j.jconrel.2018.02.020] [Citation(s) in RCA: 76] [Impact Index Per Article: 12.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/22/2017] [Revised: 02/13/2018] [Accepted: 02/14/2018] [Indexed: 12/14/2022]
Abstract
Tumor cells overexpress surface markers which are absent from normal cells. These tumor-restricted antigenic signatures are a fundamental basis for distinguishing on-target from off-target cells for ligand-directed targeting of cancer cells. Unfortunately, tumor heterogeneity impedes the establishment of a solid expression pattern for a given target marker, leading to drastic changes in quality (availability) and quantity (number) of the target. Consequently, a subset of cancer cells remains untargeted during the course of treatment, which subsequently promotes drug-resistance and cancer relapse. Since target inefficiency is only problematic for cancer treatment and not for treatment of other pathological conditions such as viral/bacterial infections, target amplification or the generation of novel targets is key to providing eligible antigenic markers for effective targeted therapy. This review summarizes the limitations of current ligand-directed targeting strategies and provides a comprehensive overview of tumor target amplification strategies, including self-amplifying systems, dual targeting, artificial markers and peptide modification. We also discuss the therapeutic and diagnostic potential of these approaches, the underlying mechanism(s) and established methodologies, mostly in the context of different nanodelivery systems, to facilitate more effective ligand-directed cancer cell monitoring and targeting.
Collapse
|